Nanomedicine approaches in acute lymphoblastic leukemia
Tài liệu tham khảo
Siegel, 2015, Cancer statistics, 2015, CA Cancer J. Clin., 65, 5, 10.3322/caac.21254
Siegel, 2012, Cancer statistics, 2012, CA Cancer J. Clin., 62, 10, 10.3322/caac.20138
Pui, 2012, Pediatric acute lymphoblastic leukemia: where are we going and how do we get there?, Blood, 120, 1165, 10.1182/blood-2012-05-378943
Loghavi, 2015, B-acute lymphoblastic leukemia/lymphoblastic lymphoma, Am. J. Clin. Pathol., 144, 393, 10.1309/AJCPAN7BH5DNYWZB
Basha, 2014, Targeted nanoparticles for pediatric leukemia therapy, Front. Oncol., 4, 10.3389/fonc.2014.00101
Park, 2014, How do we prepare ourselves for a new paradigm of medicine to advance the treatment of pediatric acute lymphoblastic leukemia?, Blood Res., 49, 3, 10.5045/br.2014.49.1.3
Napper, 2013, Targeted drug discovery for pediatric leukemia, Front. Oncol., 3, 10.3389/fonc.2013.00170
Moriyama, 2015, Inherited genetic variation in childhood acute lymphoblastic leukemia, Blood, 125, 3988, 10.1182/blood-2014-12-580001
Krishnan, 2014, Clinical nanomedicine: a solution to the chemotherapy conundrum in pediatric leukemia therapy, Clin. Pharmacol. Ther., 95, 168, 10.1038/clpt.2013.174
Pui, 2007, New therapeutic strategies for the treatment of acute lymphoblastic leukaemia, Nat. Rev. Drug Discov., 6, 149, 10.1038/nrd2240
Ness, 2015, Energy balance and fitness in adult survivors of childhood acute lymphoblastic leukemia, Blood, 125, 3411, 10.1182/blood-2015-01-621680
Turner, 2013, Effect of treatment for paediatric cancers on balance: what do we know? A review of the evidence: effect of treatment for paediatric cancers on balance, Eur. J. Cancer Care (Engl.), 22, 3, 10.1111/ecc.12019
Gocha Marchese, 2003, Strength and functional mobility in children with acute lymphoblastic leukemia, Med. Pediatr. Oncol., 40, 230, 10.1002/mpo.10266
McDougall, 2009, Quality of life in survivors of childhood cancer: a systematic review of the literature (2001–2008), Support Care Cancer, 17, 1231, 10.1007/s00520-009-0660-0
Wilson, 2014, Decline in physical activity level in the childhood cancer survivor study cohort, Cancer Epidemiol. Biomark. Prev., 23, 1619, 10.1158/1055-9965.EPI-14-0213
Carol, 2013, The anti-CD19 antibody-drug conjugate SAR3419 prevents hematolymphoid relapse postinduction therapy in preclinical models of pediatric acute lymphoblastic leukemia, Clin. Cancer Res., 19, 1795, 10.1158/1078-0432.CCR-12-3613
Mashkani, 2016
Gokbuget, 2004, Treatment with monoclonal antibodies in acute lymphoblastic leukemia: current knowledge and future prospects, Ann. Hematol., 83, 201, 10.1007/s00277-003-0752-8
2007
“Public — ETP Nanomedicine.” Online]. Available: http://www.etp-nanomedicine.eu/public. [Accessed: 11-Mar-2016.
Link, 2003, Optical properties and ultrafast dynamics of metallic nanocrystals, Annu. Rev. Phys. Chem, 54, 331, 10.1146/annurev.physchem.54.011002.103759
Hagfeldt, 2000, Molecular photovoltaics, Acc. Chem. Res, 33, 269, 10.1021/ar980112j
Kelly, 2003, The optical properties of metal nanoparticles: the influence of size, shape, and dielectric environment, J. Phys. Chem. B, 107, 668, 10.1021/jp026731y
Hao, 2004, Synthesis and optical properties of anisotropic metal nanoparticles, J. Fluoresc, 14, 331, 10.1023/B:JOFL.0000031815.71450.74
von Maltzahn, 2009, SERS-coded gold nanorods as a multifunctional platform for densely multiplexed near-infrared imaging and photothermal heating, Adv. Mater. Deerfield Beach Fla, 21, 3175, 10.1002/adma.200803464
Kneipp, 2010, Novel optical nanosensors for probing and imaging live cells, Nanomedicine Nanotechnol. Biol. Med, 6, 214, 10.1016/j.nano.2009.07.009
Boca, 2011, Flower-shaped gold nanoparticles: synthesis, characterization and their application as SERS-active tags inside living cells, Nanotechnology, 22, 55702, 10.1088/0957-4484/22/5/055702
Tong, 2009, Gold nanorods as contrast agents for biological imaging: optical properties, surface conjugation and photothermal effects, Photochem. Photobiol, 85, 21, 10.1111/j.1751-1097.2008.00507.x
Boca, 2011, Chitosan-coated triangular silver nanoparticles as a novel class of biocompatible, highly effective photothermal transducers for in vitro cancer cell therapy, Cancer Lett, 311, 131, 10.1016/j.canlet.2011.06.022
Boca-Farcau, 2014, Folic acid-conjugated, SERS-labeled silver nanotriangles for multimodal detection and targeted photothermal treatment on human ovarian cancer cells, Mol. Pharm, 11, 391, 10.1021/mp400300m
Liu, 2013, Gold nanorods coated with a thermo-responsive poly(ethylene glycol)-b-poly(N-vinylcaprolactam) corona as drug delivery systems for remotely near infrared-triggered release, Polym. Chem, 5, 799, 10.1039/C3PY01057K
Zhao, 2009, Rapid synthesis of DNA-functionalized gold nanoparticles in salt solution using mononucleotide-mediated conjugation, Bioconjug. Chem, 20, 1218, 10.1021/bc900080p
Arruebo, 2009, Antibody-conjugated nanoparticles for biomedical applications, antibody-conjugated nanoparticles for biomedical applications, J. Nanomater., 2009, e439389, 10.1155/2009/439389
Simon, 2015, Design of FLT3 inhibitor - gold nanoparticle conjugates as potential therapeutic agents for the treatment of acute myeloid leukemia, Nanoscale Res. Lett, 10, 1, 10.1186/s11671-015-1154-2
Tomuleasa, 2016, Gold nanoparticles enhance the effect of tyrosine kinase inhibitors in acute myeloid leukemia therapy, Int. J. Nanomedicine, 641, 10.2147/IJN.S94064
Chiaretti, 2014, Diagnosis and subclassification of acute lymphoblastic leukemia, Mediterr. J. Hematol. Infect. Dis, 6, 2014073, 10.4084/mjhid.2014.073
Inaba, 2013, Acute lymphoblastic leukaemia, Lancet, 381, 1943, 10.1016/S0140-6736(12)62187-4
Moorman, 2013, Risk-directed treatment intensification significantly reduces the risk of relapse among children and adolescents with acute lymphoblastic leukemia and intrachromosomal amplification of chromosome 21: a comparison of the MRC ALL97/99 and UKALL2003 trials, J. Clin. Oncol, 31, 3389, 10.1200/JCO.2013.48.9377
Pui, 2011, Biology, risk stratification, and therapy of pediatric acute leukemias: an update, J. Clin. Oncol, 29, 551, 10.1200/JCO.2010.30.7405
Woo, 2014, Childhood B-acute lymphoblastic leukemia: a genetic update, Exp. Hematol. Oncol, 3, 16, 10.1186/2162-3619-3-16
Mullighan, 2012, Molecular genetics of B-precursor acute lymphoblastic leukemia, J. Clin. Invest, 122, 3407, 10.1172/JCI61203
Krishnan, 2014, Clinical nanomedicine: a solution to the chemotherapy conundrum in pediatric leukemia therapy, Clin. Pharmacol. Ther, 95, 168, 10.1038/clpt.2013.174
Biondi, 2013, Novel clinical trials for pediatric leukemias: lessons learned from genomic analyses, Hematology, 2013, 612, 10.1182/asheducation-2013.1.612
Kang, 2010, Gene expression classifiers for relapse-free survival and minimal residual disease improve risk classification and outcome prediction in pediatric B-precursor acute lymphoblastic leukemia, Blood, 115, 1394, 10.1182/blood-2009-05-218560
Pollock, 2000, Racial differences in the survival of childhood B-precursor acute lymphoblastic leukemia: a pediatric oncology group study, J. Clin. Oncol, 18, 813, 10.1200/JCO.2000.18.4.813
Bhatia, 2002, Racial and ethnic differences in survival of children with acute lymphoblastic leukemia, Blood, 100, 1957, 10.1182/blood-2002-02-0395
Raghunathan, 2013, Anti-CRLF2 antibody-armored biodegradable nanoparticles for childhood B-ALL, Part. Part. Syst. Charact, 30, 355, 10.1002/ppsc.201200125
Van Vlierberghe, 2010, PHF6 mutations in T-cell acute lymphoblastic leukemia, Nat. Genet, 42, 338, 10.1038/ng.542
Cunningham, 2007, Pharmacogenetics of acute lymphoblastic leukemia treatment response, Expert. Opin. Pharmacother., 8, 2519, 10.1517/14656566.8.15.2519
Pui, 2008, Acute lymphoblastic leukaemia, Lancet, 371, 1030, 10.1016/S0140-6736(08)60457-2
Pui, 2013, A 50-year journey to cure childhood acute lymphoblastic leukemia, Semin. Hematol, 50, 185, 10.1053/j.seminhematol.2013.06.007
You, 2015, T-lymphoblastic leukemia/lymphoma, Am. J. Clin. Pathol, 144, 411, 10.1309/AJCPMF03LVSBLHPJ
Coustan-Smith, 2009, Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia, Lancet Oncol, 10, 147, 10.1016/S1470-2045(08)70314-0
Dear, 1993, The HOX11 gene encodes a DNA-binding nuclear transcription factor belonging to a distinct family of homeobox genes, Proc. Natl. Acad. Sci, 90, 4431, 10.1073/pnas.90.10.4431
Mauvieux, 2002, High incidence of Hox11L2 expression in children with T-ALL, Leukemia, 16, 2417, 10.1038/sj.leu.2402709
Meijerink, 2010, Genetic rearrangements in relation to immunophenotype and outcome in T-cell acute lymphoblastic leukaemia, Best Pract. Res. Clin. Haematol, 23, 307, 10.1016/j.beha.2010.08.002
Seibel, 2008, Early postinduction intensification therapy improves survival for children and adolescents with high-risk acute lymphoblastic leukemia: a report from the Children's oncology group, Blood, 111, 2548, 10.1182/blood-2007-02-070342
Paulsson, 2015, The genomic landscape of high hyperdiploid childhood acute lymphoblastic leukemia, Nat. Genet, 47, 672, 10.1038/ng.3301
Salzer, 2010, Long-term results of the pediatric oncology group studies for childhood acute lymphoblastic leukemia 1984–2001: a report from the children's oncology group, Leukemia, 24, 355, 10.1038/leu.2009.261
Linka, 2013, The impact of TEL-AML1 (ETV6-RUNX1) expression in precursor B cells and implications for leukaemia using three different genome-wide screening methods, Blood Cancer J, 3, e151, 10.1038/bcj.2013.48
Wang, 1997, Yolk sac angiogenic defect and intra-embryonic apoptosis in mice lacking the Ets-related factor TEL, EMBO J, 16, 4374, 10.1093/emboj/16.14.4374
Okuda, 1996, AML1, the target of multiple chromosomal translocations in human leukemia, is essential for normal fetal liver hematopoiesis, Cell, 84, 321, 10.1016/S0092-8674(00)80986-1
Asou, 2003, The role of a runt domain transcription factor AML1/RUNX1 in leukemogenesis and its clinical implications, Crit. Rev. Oncol. Hematol, 45, 129, 10.1016/S1040-8428(02)00003-3
Nachman, 2007, Outcome of treatment in children with hypodiploid acute lymphoblastic leukemia, Blood, 110, 1112, 10.1182/blood-2006-07-038299
Harrison, 2004, Three distinct subgroups of hypodiploidy in acute lymphoblastic leukaemia, Br. J. Haematol, 125, 552, 10.1111/j.1365-2141.2004.04948.x
Stam, 2012, MLL-AF4 driven leukemogenesis: what are we missing?, Cell Res, 22, 948, 10.1038/cr.2012.16
Meyer, 2013, The MLL recombinome of acute leukemias in 2013, Leukemia, 27, 2165, 10.1038/leu.2013.135
Hasle, 2000, Risks of leukaemia and solid tumours in individuals with Down's syndrome, Lancet Lond. Engl, 355, 165, 10.1016/S0140-6736(99)05264-2
Buitenkamp, 2012, Outcome in children with Down's syndrome and acute lymphoblastic leukemia: role of IKZF1 deletions and CRLF2 aberrations, Leukemia, 26, 2204, 10.1038/leu.2012.84
Chen, 2012, Outcome modeling with CRLF2, IKZF1, JAK, and minimal residual disease in pediatric acute lymphoblastic leukemia: a children's oncology group study, Blood, 119, 3512, 10.1182/blood-2011-11-394221
Sun, 1999, Expression of dominant-negative and mutant isoforms of the antileukemic transcription factor Ikaros in infant acute lymphoblastic leukemia, Proc. Natl. Acad. Sci, 96, 680, 10.1073/pnas.96.2.680
Losdyck, 2015, Distinct acute lymphoblastic leukemia (ALL)-associated janus kinase 3 (JAK3) mutants exhibit different cytokine-receptor requirements and JAK inhibitor specificities, J. Biol. Chem, 290, 29022, 10.1074/jbc.M115.670224
Mullighan, 2011, CREBBP mutations in relapsed acute lymphoblastic leukaemia, Nature, 471, 235, 10.1038/nature09727
Chiaretti, 2013, TP53 mutations are frequent in adult acute lymphoblastic leukemia cases negative for recurrent fusion genes and correlate with poor response to induction therapy, Haematologica, 98, e59, 10.3324/haematol.2012.076786
Schafer, 2010, Promoter hypermethylation in MLL-r infant acute lymphoblastic leukemia: biology and therapeutic targeting, Blood, 115, 4798, 10.1182/blood-2009-09-243634
Shivakumar, 2008, Biologic features and treatment outcome of secondary acute lymphoblastic leukemia–a review of 101 cases, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. ESMO, 19, 1634, 10.1093/annonc/mdn182
Chen, 2007, Stability of leukemia-associated immunophenotypes in precursor B-lymphoblastic leukemia/lymphoma: a single institution experience, Am. J. Clin. Pathol., 127, 39, 10.1309/7R6MU7R9YWJBY5V4
Timis, 2012, Paraneoplastic syndromes with connective tissue involvement. ‘It's not always lupus!, J. BUON Off. J. Balk. Union Oncol., 17, 417
Hunger, 2015, Acute lymphoblastic leukemia in children, N. Engl. J. Med., 373, 1541, 10.1056/NEJMra1400972
Pui, 2015, Childhood acute lymphoblastic leukemia: progress through collaboration, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol., 33, 2938, 10.1200/JCO.2014.59.1636
van Dongen, 2015, Minimal residual disease diagnostics in acute lymphoblastic leukemia: need for sensitive, fast, and standardized technologies, Blood, 125, 3996, 10.1182/blood-2015-03-580027
Hunger, 2015, Redefining ALL classification: toward detecting high-risk ALL and implementing precision medicine, Blood, 125, 3977, 10.1182/blood-2015-02-580043
Roberts, 2015, Genomics in acute lymphoblastic leukaemia: insights and treatment implications, Nat. Rev. Clin. Oncol., 12, 344, 10.1038/nrclinonc.2015.38
Wang, 2015, Novel management options for adult patients with progressive acute lymphoblastic leukemia: introduction, Clin. Adv. Hematol. Oncol. HO, 13, 8
Frey, 2015, How I treat adults with relapsed or refractory Philadelphia chromosome-negative acute lymphoblastic leukemia, Blood, 126, 589, 10.1182/blood-2014-09-551937
Shouval, 2015, Prediction of allogeneic hematopoietic stem-cell transplantation mortality 100days after transplantation using a machine learning algorithm: a european group for blood and marrow transplantation acute leukemia working party retrospective data mining study, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol., 33, 3144, 10.1200/JCO.2014.59.1339
Couturier, 2015, Cerebral venous thrombosis in adult patients with acute lymphoblastic leukemia or lymphoblastic lymphoma during induction chemotherapy with l-asparaginase: The GRAALL experience, Am. J. Hematol., 90, 986, 10.1002/ajh.24130
Shalabi, 2015, Beyond CD19: opportunities for future development of targeted immunotherapy in pediatric relapsed-refractory acute leukemia, Front. Pediatr., 3, 10.3389/fped.2015.00080
Vedi, 2014, Antibody therapy for pediatric leukemia, Pediatr. Oncol., 4, 82
Hoelzer, 2011, Novel antibody-based therapies for acute lymphoblastic leukemia, Hematol. Educ. Program Am. Soc. Hematol. Am. Soc. Hematol. Educ. Program, 2011, 243, 10.1182/asheducation-2011.1.243
Raponi, 2011, Flow cytometric study of potential target antigens (CD19, CD20, CD22, CD33) for antibody-based immunotherapy in acute lymphoblastic leukemia: analysis of 552 cases, Leuk. Lymphoma, 52, 1098, 10.3109/10428194.2011.559668
Annesley, 2015, Novel agents for the treatment of childhood acute leukemia, Ther. Adv. Hematol., 6, 61, 10.1177/2040620714565963
Chan, 1988, A simple guide to the terminology and application of leucocyte monoclonal antibodies, Histopathology, 12, 461, 10.1111/j.1365-2559.1988.tb01967.x
Schlossman, 1994, CD antigens 1993, J. Immunol. Baltim. Md 1950, 152, 1
1997, Bull. World Health Organ., 75, 385
Zola, 2003, Human leucocyte differentiation antigen nomenclature: update on CD nomenclature. Report of IUIS/WHO Subcommittee, J. Immunol. Methods, 275, 1
Zola, 2005, CD molecules 2005: human cell differentiation molecules, Blood, 106, 3123, 10.1182/blood-2005-03-1338
Zola, 2007, CD molecules 2006–human cell differentiation molecules, J. Immunol. Methods, 319, 1, 10.1016/j.jim.2006.11.001
“cdlist.txt.” [Online]. Available: http://www.uniprot.org/docs/cdlist. [Accessed: 11-Mar-2016].
Ginaldi, 1996, Differential expression of CD3 and CD7 in T-cell malignancies: a quantitative study by flow cytometry, Br. J. Haematol., 93, 921, 10.1046/j.1365-2141.1996.d01-1720.x
Leong, 2003
Smith, 1996, Ten years of Orthoclone OKT3 (muromonab-CD3): a review, J. Transpl. Coord. Off. Publ. North Am. Transpl. Coord. Organ. NATCO, 6, 109
Gramatzki, 1995, Therapy with OKT3 monoclonal antibody in refractory T cell acute lymphoblastic leukemia induces interleukin-2 responsiveness, Leukemia, 9, 382
Gunther, 1993, In vivo anti-leukemic efficacy of anti-CD7-pokeweed antiviral protein immunotoxin against human T-lineage acute lymphoblastic leukemia/lymphoma in mice with severe combined immunodeficiency, Leukemia, 7, 298
Frankel, 1997, Therapy of patients with T-cell lymphomas and leukemias using an anti-CD7 monoclonal antibody-ricin A chain immunotoxin, Leuk. Lymphoma, 26, 287, 10.3109/10428199709051778
Galy, 1995, Human T, B, natural killer, and dendritic cells arise from a common bone marrow progenitor cell subset, Immunity, 3, 459, 10.1016/1074-7613(95)90175-2
Uckun, 1999, Treatment of therapy-refractory B-lineage acute lymphoblastic leukemia with an apoptosis-inducing CD19-directed tyrosine kinase inhibitor, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res., 5, 3906
Breton, 2014, A novel anti-CD19 monoclonal antibody (GBR 401) with high killing activity against B cell malignancies, J. Hematol. Oncol., 7, 33, 10.1186/1756-8722-7-33
Löffler, 2000, A recombinant bispecific single-chain antibody, CD19 x CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocytes, Blood, 95, 2098, 10.1182/blood.V95.6.2098
Topp, 2011, Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival, J. Clin. Oncol., 29, 2493, 10.1200/JCO.2010.32.7270
Ceppi, 2016, Safety and efficacy of intrathecal rituximab in children with B cell lymphoid CD20+ malignancies: an international retrospective study: safety and efficacy of intrathecal rituximab in children, Am. J. Hematol., 10.1002/ajh.24329
Vacchelli, 2015, Trial watch: tumor-targeting monoclonal antibodies for oncological indications, OncoImmunology, 4, e985940, 10.4161/2162402X.2014.985940
Leonard, 2007, Preclinical and clinical evaluation of epratuzumab (anti-CD22 IgG) in B-cell malignancies, Oncogene, 26, 3704, 10.1038/sj.onc.1210370
Rytting, 2014, Initial experience with CMC-544 (inotuzumab ozogamicin) in pediatric patients with relapsed B-cell acute lymphoblastic leukemia, Pediatr. Blood Cancer, 61, 369, 10.1002/pbc.24721
Waldmann, 1993, The interleukin-2 receptor: a target for monoclonal antibody treatment of human T-cell lymphotrophic virus I-induced adult T-cell leukemia, Blood, 82, 1701, 10.1182/blood.V82.6.1701.1701
Rowe, 2013, Gemtuzumab ozogamicin in acute myeloid leukemia: a remarkable saga about an active drug, Blood, 121, 4838, 10.1182/blood-2013-03-490482
Tibes, 2006, Activity of alemtuzumab in patients with CD52-positive acute leukemia, Cancer, 106, 2645, 10.1002/cncr.21901
Angiolillo, 2009, A phase II study of campath-1H in children with relapsed or refractory acute lymphoblastic leukemia: a children's oncology group report, Pediatr. Blood Cancer, 53, 978, 10.1002/pbc.22209
Gharwan, 2015, Kinase inhibitors and monoclonal antibodies in oncology: clinical implications, Nat. Rev. Clin. Oncol., 13, 209, 10.1038/nrclinonc.2015.213
Hansel, 2010, The safety and side effects of monoclonal antibodies, Nat. Rev. Drug Discov., 9, 325, 10.1038/nrd3003
Jabbour, 2015, New insights into the pathophysiology and therapy of adult acute lymphoblastic leukemia, Cancer, 121, 2517, 10.1002/cncr.29383
Chiaretti, 2015, Management of adult Ph-positive acute lymphoblastic leukemia, Hematol. Educ. Program Am. Soc., 2015, 406, 10.1182/asheducation-2015.1.406
Curran, 2015, How I treat acute lymphoblastic leukemia in older adolescents and young adults, Blood, 125, 3702, 10.1182/blood-2014-11-551481
Yeh, 2012, Gold nanoparticles: preparation, properties, and applications in bionanotechnology, Nanoscale, 4, 1871, 10.1039/C1NR11188D
Alonso, 2004, Nanomedicines for overcoming biological barriers, Biomed. Pharmacother., 58, 168, 10.1016/j.biopha.2004.01.007
Fu, 2001, Size effects on the optical properties of organic nanoparticles, J. Am. Chem. Soc., 123, 1434, 10.1021/ja0026298
Scholes, 2008, Controlling the optical properties of inorganic nanoparticles, Adv. Funct. Mater., 18, 1157, 10.1002/adfm.200800151
Pellegrino, 2005, On the development of colloidal nanoparticles towards multifunctional structures and their possible use for biological applications, Small, 1, 48, 10.1002/smll.200400071
Sailor, 2012, Hybrid nanoparticles for detection and treatment of cancer, Adv. Mater, 24, 3779, 10.1002/adma.201200653
Lee, 2011, Multifunctional mesoporous silica nanocomposite nanoparticles for theranostic applications, Acc. Chem. Res., 44, 893, 10.1021/ar2000259
Tang, 2013, Nonporous silica nanoparticles for nanomedicine application, Nano Today, 8, 290, 10.1016/j.nantod.2013.04.007
Ruoslahti, 2010, Targeting of drugs and nanoparticles to tumors, J. Cell Biol., 188, 759, 10.1083/jcb.200910104
Huang, 2009, Aptamer-modified gold nanoparticles for targeting breast cancer cells through light scattering, J. Nanopart. Res., 11, 775, 10.1007/s11051-008-9424-x
Sensenig, 2012, Magnetic nanoparticle-based approaches to locally target therapy and enhance tissue regeneration in vivo, Nanomed., 7, 1425, 10.2217/nnm.12.109
Torchilin, 2011, Tumor delivery of macromolecular drugs based on the EPR effect, Adv. Drug Deliv. Rev., 63, 131, 10.1016/j.addr.2010.03.011
Muthu, 2013, Theranostic liposomes for cancer diagnosis and treatment: current development and pre-clinical success, Expert Opin. Drug Deliv., 10, 151, 10.1517/17425247.2013.729576
Farokhzad, 2009, Impact of nanotechnology on drug delivery, ACS Nano, 3, 16, 10.1021/nn900002m
Gao, 2010, pH-responsive nanoparticles for drug delivery, Mol. Pharm., 7, 1913, 10.1021/mp100253e
Schmaljohann, 2006, Thermo- and pH-responsive polymers in drug delivery, Adv. Drug Deliv. Rev., 58, 1655, 10.1016/j.addr.2006.09.020
Bibi, 2012, Trigger release liposome systems: local and remote controlled delivery?, J. Microencapsul., 29, 262, 10.3109/02652048.2011.646330
Ganta, 2008, A review of stimuli-responsive nanocarriers for drug and gene delivery, J. Control. Release, 126, 187, 10.1016/j.jconrel.2007.12.017
Gong, 2013, Thermosensitive polymeric hydrogels as drug delivery systems, Curr. Med. Chem., 20, 79, 10.2174/0929867311302010009
Kneidl, 2014, Thermosensitive liposomal drug delivery systems: state of the art review, Int. J. Nanomedicine, 9, 4387
Li, 2013, Mild hyperthermia triggered doxorubicin release from optimized stealth thermosensitive liposomes improves intratumoral drug delivery and efficacy, J. Control. Release, 168, 142, 10.1016/j.jconrel.2013.03.011
Qian, 2013, Thermosensitive PNIPAM semi-hollow spheres for controlled drug release, J. Mater. Chem. B, 1, 3464, 10.1039/c3tb20527d
Yang, 2014, Advances in self-assembled chitosan nanomaterials for drug delivery, Biotechnol. Adv., 32, 1301, 10.1016/j.biotechadv.2014.07.007
Rahikkala, 2015, Thermoresponsive nanoparticles of self-assembled block copolymers as potential carriers for drug delivery and diagnostics, Biomacromolecules, 16, 2750, 10.1021/acs.biomac.5b00690
Latorre, 2014, Glutathione-triggered drug release from nanostructures, Curr. Top. Med. Chem., 14, 2662, 10.2174/1568026614666141215143659
Schattling, 2014, Multi-stimuli responsive polymers – the all-in-one talents, Polym. Chem., 5, 25, 10.1039/C3PY00880K
Zhang, 2015, Polymer-coated hollow mesoporous silica nanoparticles for triple-responsive drug delivery, ACS Appl. Mater. Interfaces, 7, 18179, 10.1021/acsami.5b05893
Zhang, 2015, Multi-stimuli-responsive magnetic assemblies as tunable releasing carriers, J. Mater. Chem. B, 3, 6026, 10.1039/C5TB00845J
Dinauer, 2005, Selective targeting of antibody-conjugated nanoparticles to leukemic cells and primary T-lymphocytes, Biomaterials, 26, 5898, 10.1016/j.biomaterials.2005.02.038
Fan, 2015, A polydopamine nanosphere based highly sensitive and selective aptamer cytosensor with enzyme amplification, Chem. Commun., 52, 406, 10.1039/C5CC06754E
Pressly, 2013, Nanoparticle PET/CT imaging of natriuretic peptide clearance receptor in prostate cancer, Bioconjug. Chem., 24, 196, 10.1021/bc300473x
Gui, 2014, Intracellular fluorescent thermometry and photothermal-triggered drug release developed from gold nanoclusters and doxorubicin dual-loaded liposomes, Chem. Commun., 50, 1546, 10.1039/c3cc47981a
Batrakova, 2008, Pluronic block copolymers: evolution of drug delivery concept from inert nanocarriers to biological response modifiers, J. Control. Release, 130, 98, 10.1016/j.jconrel.2008.04.013
Krishnan, 2013, Dexamethasone-loaded block copolymer nanoparticles induce leukemia cell death and enhance therapeutic efficacy: a novel application in pediatric nanomedicine, Mol. Pharm., 10, 2199, 10.1021/mp300350e
Krishnan, 2015, CD19-targeted Nanodelivery of doxorubicin enhances therapeutic efficacy in B-cell acute lymphoblastic leukemia, Mol. Pharm, 12, 2101, 10.1021/acs.molpharmaceut.5b00071
Allen, 2013, Liposomal drug delivery systems: from concept to clinical applications, Adv. Drug Deliv. Rev., 65, 36, 10.1016/j.addr.2012.09.037
Silverman, 2013, Marqibo® (vincristine sulfate liposome injection) improves the pharmacokinetics and pharmacodynamics of vincristine, Cancer Chemother. Pharmacol., 71, 555, 10.1007/s00280-012-2042-4
Gulati, 1998, Lipophilic drug derivatives in liposomes, Int. J. Pharm., 165, 129, 10.1016/S0378-5173(98)00006-4
Immordino, 2006, Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential, Int. J. Nanomedicine, 1, 297
Kobayashi, 1975, Enhancement of the cancer chemotherapeutic effect of cytosine arabinoside entrapped in liposomes on mouse leukemia L-1210, Gann Gan, 66, 719
Embree, 1997, Validation of a high-performance liquid chromatographic assay method for quantification of total vincristine sulfate in human plasma following administration of vincristine sulfate liposome injection, J. Pharm. Biomed. Anal., 16, 675, 10.1016/S0731-7085(97)00087-3
Embree, 1998, Pharmacokinetic behavior of vincristine sulfate following administration of vincristine sulfate liposome injection, Cancer Chemother. Pharmacol., 41, 347, 10.1007/s002800050750
“FDA-Approved Marqibo® (vinCRIStine sulfate LIPOSOME injection) www.MARQIBO.com.” [Online]. Available: http://www.marqibo.com/. [Accessed: 30-Jun-2016].
Moore, 2013, Vincristine sulfate liposomal injection for acute lymphoblastic leukemia, Int. J. Nanomedicine, 4361, 10.2147/IJN.S54657
“Paper: Marqibo®, Vincristine Sulfate Liposome Injection, for the Treatment of Advanced, Relapsed or Refractory Philadelphia Chromosome-Negative (Ph−) Acute Lymphoblastic Leukemia (ALL) in an Adolescent Young Adult (AYA) Population.” [Online]. Available: https://ash.confex.com/ash/2015/webprogram/Paper87149.html. [Accessed: 30-Jun-2016].
Shah, 2016, Vincristine sulfate liposomes injection (VSLI, Marqibo®): results from a phase I study in children, adolescents, and young adults with refractory solid tumors or leukemias: pediatric phase I Marqibo®, Pediatr. Blood Cancer, 63, 997, 10.1002/pbc.25937
Carol, 2015, Efficacy of CPX-351, (cytarabine:daunorubicin) liposome injection, against acute lymphoblastic leukemia (ALL) xenograft models of the pediatric preclinical testing program: CPX-351 in acute lymphoblastic leukemia, Pediatr. Blood Cancer, 62, 65, 10.1002/pbc.25133
Myers, 2014, CD19-antigen specific nanoscale liposomal formulation of a SYK P-site inhibitor causes apoptotic destruction of human B-precursor leukemia cells, Integr. Biol., 6, 766, 10.1039/C4IB00095A
Bhushan, 2016, The augmented anticancer potential of AP9-cd loaded solid lipid nanoparticles in human leukemia molt-4 cells and experimental tumor, Chem. Biol. Interact., 244, 84, 10.1016/j.cbi.2015.11.022
Liu, 2013, Graphene and graphene oxide as new nanocarriers for drug delivery applications, Acta Biomater., 9, 9243, 10.1016/j.actbio.2013.08.016
Omidi, 2011
Bao, 2011, Chitosan-functionalized graphene oxide as a nanocarrier for drug and gene delivery, Small, 7, 1569, 10.1002/smll.201100191
Chen, 2014, One-step reduction and PEGylation of graphene oxide for photothermally controlled drug delivery, Biomaterials, 35, 4986, 10.1016/j.biomaterials.2014.02.032
You, 2015, Graphene oxide-based nanocarriers for cancer imaging and drug delivery, Curr. Pharm. Des, 21, 3215, 10.2174/1381612821666150531170832
Akhavan, 2014, Spongy graphene electrode in electrochemical detection of leukemia at single-cell levels, Carbon, 79, 654, 10.1016/j.carbon.2014.08.058
Akhavan, 2014, Ultra-sensitive detection of leukemia by graphene, Nanoscale, 6, 14810, 10.1039/C4NR04589K
Zoref-Shani, 1995, Metabolism of guanine and guanine nucleotides in primary rat cardiomyocyte cultures, Biochem. Mol. Med., 55, 149, 10.1006/bmme.1995.1045
Ladj, 2013, Individual inorganic nanoparticles: preparation, functionalization and in vitro biomedical diagnostic applications, J. Mater. Chem. B, 1, 1381, 10.1039/c2tb00301e
Gerhards, 2008, Conjugating luminescent CdTe quantum dots with biomolecules†, J. Phys. Chem. B, 112, 14482, 10.1021/jp8030094
van Vlerken, 2007, Poly(ethylene glycol)-modified nanocarriers for tumor-targeted and intracellular delivery, Pharm. Res., 24, 1405, 10.1007/s11095-007-9284-6
Tan, 2009, Inorganic nanoparticles for biomedical applications, 272
Anker, 2008, Biosensing with plasmonic nanosensors, Nat. Mater., 7, 442, 10.1038/nmat2162
Han, 2007, Functionalized gold nanoparticles for drug delivery, Nanomed., 2, 113, 10.2217/17435889.2.1.113
Yan, 2015, A label-free immunosensor for detecting common acute lymphoblastic leukemia antigen (CD10) based on gold nanoparticles by quartz crystal microbalance, Sens. Actuators B Chem., 210, 248, 10.1016/j.snb.2014.12.104
Nguyen, 2010, Detection of chronic lymphocytic leukemia cell surface markers using surface enhanced Raman scattering gold nanoparticles, Cancer Lett., 292, 91, 10.1016/j.canlet.2009.11.011
Danesh, 2015, Targeted and controlled release delivery of daunorubicin to T-cell acute lymphoblastic leukemia by aptamer-modified gold nanoparticles, Int. J. Pharm., 489, 311, 10.1016/j.ijpharm.2015.04.072
Liu, 2015, Targeted intracellular controlled drug delivery and tumor therapy through in situ forming Ag nanogates on mesoporous silica nanocontainers, ACS Appl. Mater. Interfaces, 7, 11930, 10.1021/acsami.5b01787
Zhou, 2016, Hyaluronic acid-coated magnetic nanoparticles-based selective collection and detection of leukemia cells with quartz crystal microbalance, Sens. Actuators B Chem., 223, 9, 10.1016/j.snb.2015.09.063
Fadeel, 2010, Better safe than sorry: understanding the toxicological properties of inorganic nanoparticles manufactured for biomedical applications, Adv. Drug Deliv. Rev., 62, 362, 10.1016/j.addr.2009.11.008
Liu, 2003, Gold-cluster degradation by the transition of B-DNA into A-DNA and the formation of nanowires, Angew. Chem. Int. Ed., 42, 2853, 10.1002/anie.200250235
Semmler-Behnke, 2008, Biodistribution of 1.4- and 18-nm gold particles in rats, Small, 4, 2108, 10.1002/smll.200800922
Pernodet, 2006, Adverse effects of citrate/gold nanoparticles on human dermal fibroblasts, Small, 2, 766, 10.1002/smll.200500492
Balasubramanian, 2010, Biodistribution of gold nanoparticles and gene expression changes in the liver and spleen after intravenous administration in rats, Biomaterials, 31, 2034, 10.1016/j.biomaterials.2009.11.079
Jia, 2009, Potential oxidative stress of gold nanoparticles by induced-NO releasing in serum, J. Am. Chem. Soc., 131, 40, 10.1021/ja808033w
Li, 2008, Gold nanoparticles induce oxidative damage in lung fibroblasts in vitro, Adv. Mater., 20, 138, 10.1002/adma.200701853
Khlebtsov, 2011, Biodistribution and toxicity of engineered gold nanoparticles: a review of in vitro and in vivo studies, Chem. Soc. Rev., 40, 1647, 10.1039/C0CS00018C
De Jong, 2008, Particle size-dependent organ distribution of gold nanoparticles after intravenous administration, Biomaterials, 29, 1912, 10.1016/j.biomaterials.2007.12.037
Wiwanitkit, 2009, Effect of gold nanoparticles on spermatozoa: the first world report, Fertil. Steril., 91, e7, 10.1016/j.fertnstert.2007.08.021
Kim, 2009, Intravenously administered gold nanoparticles pass through the blood–retinal barrier depending on the particle size, and induce no retinal toxicity, Nanotechnology, 20, 505101, 10.1088/0957-4484/20/50/505101
Wang, 2010, Characterization of gold nanorods in vivo by integrated analytical techniques: their uptake, retention, and chemical forms, Anal. Bioanal. Chem., 396, 1105, 10.1007/s00216-009-3302-y
Sonavane, 2008, Biodistribution of colloidal gold nanoparticles after intravenous administration: effect of particle size, Colloids Surf. B Biointerfaces, 66, 274, 10.1016/j.colsurfb.2008.07.004
Zhang, 2009, Influence of anchoring ligands and particle size on the colloidal stability and in vivo biodistribution of polyethylene glycol-coated gold nanoparticles in tumor-xenografted mice, Biomaterials, 30, 1928, 10.1016/j.biomaterials.2008.12.038
Chen, 2009, Assessment of the in vivo toxicity of gold nanoparticles, Nanoscale Res. Lett., 4, 858, 10.1007/s11671-009-9334-6
Lasagna-Reeves, 2010, Bioaccumulation and toxicity of gold nanoparticles after repeated administration in mice, Biochem. Biophys. Res. Commun., 393, 649, 10.1016/j.bbrc.2010.02.046